用户名: 密码: 验证码:
乙酰甲喹临床前毒理学研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
从二十世纪七十年代开始,喹噁啉1,4-氮氧化衍生物被认为是具有广泛生物活性的化学物质。它们被广泛用于促进动物生长和提高饲料转化率。卡巴氧和喹乙醇引起肝毒性,肾上腺毒性,遗传毒性,光敏毒性,生长繁殖毒性以及致癌遗传毒性,已经被禁用或者限制使用在动物饲料中。乙酰甲喹是新一代的喹嗯啉类衍生物,被中国允许使用于家禽和家畜。然而,根据我们的了解,对乙酰甲喹的毒性并没有进行深入的研究。为了研究乙酰甲喹的毒性,进行临床前毒理试验,以便能对乙酰甲喹进行安全评估。
     1遗传毒性试验
     1.1细菌回复突变试验/Ames实验。用6株鼠伤寒沙门氏菌(TA97,TA98,TA100, TA102, TA1535,和TA1537)以及S9检测乙酰甲喹的基因突变特性。S9用苯巴比妥钠和p-萘黄酮多次诱导大鼠获得。乙酰甲喹剂量为1,2.6,6.9,18.2和50μg/板。TA97和TA 1535测试菌株(不加S9),浓度≥1 pg/ml;TA98,TA100和TA1537浓度≥2.6 pg/ml;TA102,浓度≥6.9μg/ml,不加鼠肝S9,可以观察到明显的基因突变现象。
     1.2染色体畸变实验为研究乙酰甲喹的潜在致突变性,对V79细胞进行染色体畸变分析。乙酰甲喹5个不同的浓度(0,1.25,2.5,5和10 pg/ml)。结果显示:加或不加肝S9均可诱导V79染色体畸变。
     1.3体内微核试验将昆明小白鼠暴露于乙酰甲喹药物作用下,观察骨髓嗜多然红细胞,检测诱导微核产生的能力。以单一剂量0,5,50和500 mg/kg b.w.分别给予8只雌性小白鼠和8只雄性小白鼠。结果显示:以500 mg/kg b.w的剂量给予的雌雄小鼠出现显著的微核数量。
     2急性毒性试验采用OECD指南425规定的Up and Down procedure (UDP)法,用雌雄Wistar大鼠,研究乙酰甲喹的LD50值。乙酰甲喹溶于0.5%的硝酸纤维素钠中,口服给予单一剂量175,550或2000 mg/kg b.w。观察14d,通过AOT425统计方案评估LD50值。结果显示:Wistar大鼠的LD50为550mg/kg b.w.,昆明小白鼠LD50为988.4 mg/kg b.w.。
     3临床前90d喂养实验为研究乙酰甲喹的临川前毒性,以0,55,110和275mg/kg的剂量饲喂Wistar大鼠,275 mg/kg的剂量组大鼠出现明显体重下降。90 d后进行尸检,发现给予275 mg/kg剂量的雄性大鼠肾脏重量明显减小,而雌性肝脏和肾上腺增加。雄性大鼠丙氨酸转移酶,丙二醛浓度,雌性大鼠的超氧化物歧化酶,天冬氨酸氨基转移酶增加。275 mg/kg剂量下,还可以观察到雌雄大鼠显著的钠,钾水平下降。我们得出结论,乙酰甲喹能导致肝脏和肾上腺组织学变化,以及血清学上的诸多改变。
     4鼠的喂养致畸试验和两代繁殖试验根据目前FDA指南,研究乙酰甲喹的潜在的两代繁殖毒性以及致畸毒性。Wistar大鼠雌雄各25只,浓度剂量为0,25,55,110和275 mg/kg。F0代大鼠给药12周,在交配前,交配,妊娠,哺乳一直到死亡。F1代大鼠断奶后以同样方式给药。F2代维持21d的泌乳期,然后处死。F1代再次交配观察致畸毒性。临床观察,体重和饲料消耗率。乙酰甲喹275 mg/kg的剂量组发现F0和F1代雌性大鼠主要的观察指标体重,卵巢和肾上腺重量明显比对照组降低。275 mg/kg,的剂量组肝,肾上腺和睾丸出现萎缩。致畸毒性试验中胚胎重量下降,乙酰甲喹具有胚胎毒性。
     5长期、慢性毒性试验
     5.1长期毒性试验研究雄性大鼠肾上腺剂量依赖性长期毒性。大鼠以5个不同的剂量(0,25,55,110和275 mg/kg)饲喂乙酰甲喹180 d。观察到实验组血浆钾离子水平明显比对照组出现明显的剂量依赖性增加。高剂量绍(110,275 mg/kg)发现血浆钠离子水平下降,皮质酮和醛固酮释放减少。而且275mg/kg剂量组的肾上腺出现明显的组织损伤。为更深一步研究乙酰甲喹的肾上腺毒性机理,检测血液中氧化指标和肾上腺中5个重要酶醛固酮和皮质酮基因的mRNA水平。110,275 mg/kg剂量组,谷胱甘肽还原酶和超氧化物歧化酶减少。同样的剂量下,乙酰甲喹下调CYP11A1,CYP11B1和CYP11B2基因的mRNA表达,上调位于内置网上的CYP21和3β-HSD基因表达。结论:乙酰甲喹对雄性大鼠肾上腺造成剂量依赖性长期毒性,因此提示乙酰甲喹对动物和消费者的毒性,机理可能涉及到氧化应激和类固醇合成。
     5.2长期睾丸毒性试验该实验设计研究乙酰甲喹通过氧化应激和甾体基因表达引起的睾丸毒性,以及睾丸毒性机理。成年雄性大鼠分别饲喂不同浓度的的乙酰甲喹(0,25,55,110和275 mg/kg),180d。和对照组相比,剂量组110和275 mg/kg的SOD,GSH和(8-OHdG)明显上高,仅275 mg/kg剂量组MDA轻微上升。更近一步研究,用LC/MS-IT-TOF分析,发现代谢物M11。275 mg/kg剂量组体重,睾丸重量,睾酮以及110和275 mg/kg剂量组的血清卵泡刺激激素下降,而LH水平上升。尸检发现生殖细胞耗竭,生精小管以及睾丸收缩管内容物。和对照组相比,275 mg/kg剂量组,睾丸中StAR,P450scc和1713-HSD基因表达水平明显下降,而110 mg/kg剂量组的AR和3β-HSD mRNA表达显著上升。总之,我们第一次发现直接的证据:乙酰甲喹体内通过N→O基团还原代谢,产生自由基。这可能有助于更好的理解喹噁啉类衍生物所引起的雄性不育问题。
Since 1970s, quinoxaline 1,4-dioxide derivatives (QdNOs) have been regarded as an interesting range of biological active compounds. They are widely used in subtherapeutic levels to promote growth and improve efficiency of feed conversion in animal feed. QdNO derivatives, carbadox (CBX) and olaquindox (OLA) had raised serious health hazards of hepatotoxicity, adrenal toxicity, genotoxicity, phototoxicity, developmental and reproductive toxicity and carcinogenicity and been been prohibited or limited to use in animal feed. Mequindox (MEQ) is a novel quinoxaline derivative this family, have been approved in China for use in diet for livestock and poultry. However, according to our knowledge, a comprehensive toxicological evaluation of MEQ has not yet conducted. To investigate the toxicity of MEQ, preclinical toxicity tests were conducted to accumulate information for safety evaluation of this important agent.
     1 Genotoxicity tests
     1.1 Bacterial Reverse Mutation test/Ames test. The mutagenicity of MEQ was tested in a reverse mutation assay using six S. typhimurium strains (TA97, TA98, TA100, TA102, TA1535, and TA1537) with and without S9-mix from phenobarbital sodium andβ-Naphthoflavone induced rats using triplicate plates. The tests were conducted with doses 1,2.6,6.9,18.2 and 50μg/plate MEQ. A clear mutagenic response was seen in strains TA97 and TA 1535 (without S9) at concentrations≥1μg/ml, TA98, TA100 and TA1537 at concentrations≥2.6μg/ml, and TA102 at concentrations≥6.9μg/ml, with or without rat liver S9-mix.
     1.2 Chromosomal aberration test. To investigate the mutagenic potential of MEQ in cell cultures, chromosomal aberration test was conducted in V79 cell line. MEQ was administed to 5 different concentrations (0,1.25,2.5,5 and 10). The results had shown that there was induction of chromosome aberrations in cultured V79 cells with or without S9 at concentrations≥1.25μg/ml.
     1.3 In-vivo micronucleus assay. The test was performed to test the induction of micronuclei in polychromatic erythrocytes from femural bone marrow of Kunming mice resulting from exposure to MEQ. The MEQ was administered to groups of eight male and eight female mice at a single dose level of 0,5,50, and 500 mg/kg b.w. The results show that MEQ can cause a significant increase in the number of micronuclei at doses of 50 mg/kg b.w. in both sexes of mice.
     2 Acute toxicity test. To determine the LD50 values in female Wistar rats and female Kunming mice, Up and Down procedure (UDP) was performed according to the OECD Guideline 425. MEQ was suspended in 0.5% carboxymethyl-cellulose solution (CMC) and given as a single oral dose of 175, 550 or 2000 mg/kg body weight by gavage. The animals were monitored for up to 14 days and estimated LD50 values were calculated by AOT425 statistical program. The results showed that the LD50 value was 550 mg/kg b.w. in Wistar rats and 988.4 mg/kg b.w. in Kunming mice.
     3 subchronic 90-day feeding test in rats. To investigate the potential subchronic toxicity of MEQ, Wistar rats were fed diets containing 0,55,110 or 275 mg MEQ/kg. There was a reduction in body weight of rats fed 275 mg MEQ/kg diet. At 90 days autopsy, a significant decrease in the kidney weight was observed in males while an increase in relative liver and adrenal weights were observed in females fed 275 mg MEQ/kg diet. There was a significant increased in alanine-aminotransferase (ALT) and malondialdehyde (MDA) concentrations in males, superoxide dismutase (SOD) activities in females, and aspartate-aminotransferase (AST) levels in serum of both genders fed 275 mg MEQ/kg diet. Other toxic effects of 275 mg MEQ/kg diet included significant decrease in sodium and significant increase in potassium concentrations in serum in both genders. We may conclude that MEQ can induce hepatic and adrenal histological changes as well as leaking of different serum constituents in Wistar rats. Based on the subchronic study,110 mg/kg diet was determined to be NOAEL in both sexes of Wistar rats.
     4 Feeding teratogenic and two generation reproduction test in rats. The potential two generation reproductive toxicity and teratogenic studies for MEQ were conducted using current FDA guidelines. Groups of 25 male and 25 female Wistar rats were offered dietary concentrations of 0,25,55,110 and 275 mg/kg MEQ. Parental (F0) generation rats (4-6 weeks old) were treated for 12 weeks prior to mating, throughout mating, gestation and lactation until sacrificed. F1-generation rats were dosed similarly, beginning at weaning. The F2a generation pups were maintained through 21 days of lactation and then sacrificed. The F1-generation rats were mated again to observe teratogenicity. Clinical observations, body weights and feed consumption were recorded routinely. The mean body weight, ovary and adrenal weights in the females of F0 and F1 given 275 mg/kg MEQ were significantly suppressed than the controls. At 275 mg/kg, liver, kidneys adrenal and testis have degenerative changes. Early pup mortality was found in the high dose groups. In the teratogenic test, there was decrease in the weight of pups with kinky tail. The MEQ was found to be embryotoxic at 275 mg/kg. Based on this study,25 mg/kg diet was determined to be NOAEL in the Wistar rats.
     5. Long term/chronic toxicity test in rats.
     5.1 Long term adrenal toxicity. This study was to investigate the dose-dependent long-term toxicity in the adrenal of male rats. Rats were fed with MEQ for 180 days at five different doses (0,25,55,110 and 275 mg/kg, respectively). An increase in plasma potassium (K+) levels was observed in MEQ groups in comparison to control animals, and this increase was dose-dependent. Both high doses (110,275 mg/kg) of MEQ treatment showed a significant decline in plasma sodium (Na+), corticosterone and aldosterone release. Moreover, the 275 mg/kg treated group exhibited a marked pathological damage in adrenal tissue. To further explore the mechanism of MEQ on adrenal toxicity, oxidative indexes in blood and mRNA levels of five enzymes that are important for aldosterone and corticosterone biosynthesis were detected. Significant changes of reduced glutathione (GSH) and superoxide dismutase (SOD) in plasma were observed in the group treated at high doses (110,275 mg/kg). At the same doses, MEQ treatment down- regulated the mRNA levels of CYP11A1, CYP11B1 and CYP11B2 which located in mitochondria, but up-regulated mRNA levels of CYP21 and 3β-HSD which located in endoplasmic reticulum. In conclusion, we reported the dose-dependent long-term toxicity of MEQ on adrenal gland in male rats, which raise awareness of its toxic effects to animals and consumers, and its mechanism may involve in oxidative stress and steroid hormone biosynthesis pathway.
     5.2 Long term testicular toxicity. This study was designed to investigate the hypothesis that MEQ exerts testicular toxicity by causing oxidative stress and steroidal gene expression profiles and determine mechanism of MEQ testicular toxicity. In this study, adult male Wistar rats were fed with MEQ for 180 days at five different doses as 0,25,55,110 and 275 mg/kg, respectively. In comparison to control, SOD, GSH and 8-hydroxydeoxyguanosine (8-OHdG) levels were elevated at 110 and 275 mg/kg MEQ, whereas the MDA level was slightly increase at only 275 mg/kg. Furthermore, in LC/MS-IT-TOF analysis, one metabolite 2-isoethanol 4-desoxymequindox (M11) was found in the testis. There was significant decrease in body weight, testicular weight and testosterone at 275 mg/kg, serum follicular stimulating hormone (FSH) at 110 and 275 mg/kg, while lutinizing hormone (LH) levels were elevated at 110 mg/kg. Moreover, histopathology of testis exhibited germ cell depletion, contraction of seminiferous tubules and disorganization of the tubular contents of testis. Compared with control, mRNA expression of StAR, P450scc and 17β-HSD in testis was significantly decreased after exposure of 275 mg/kg MEQ while AR and 3P-HSD mRNA expression were significantly elevated at the 110 mg/kg MEQ group. Taken together, our findings provide the first and direct evidence in vivo for the formation of free radicals during the MEQ metabolism through N→O group reduction, which may have implications to understand the possible mechanism of male infertility related to quinoxaline derivatives.
引文
Abd-Allah, A.R., Aly, H.A., Moustafa, A.M., Abdel-Aziz, A.A., Hamada, F.M., 2000. Adverse testicular effects of some quinolone members in rats. Pharmacological Research 41,211-219.
    Aitken, R.J., Baker, M.A.,2004. Oxidative stress and male reproductive biology. Reproduction, Fertility and Development 16,581-588.
    Aitken, R.J., Baker, M.A.,2006. Oxidative stress, sperm survival and fertility control. Molecular and Cell Endocrinology 250,66-69.
    Aitken, R.J., Ryan, A.L., Baker, M.A., McLaughlin, E.A.,2004. Redox activity associated with the maturation and capacitation of mammalian spermatozoa. Free Radical Biology and Medicine 36,994-1010.
    Ajani, A.E., Marwick, T.H., Krum, H.,2006. Aldosterone antagonists:a new treatment option for patients with post-myocardial infarction heart failure. Cardiovascular Revascularization Medicine 4,234-236.
    Arimoto, S., Negishi, T., Hayatsu, H.,1980. Inhibitory effect of hemin on the mutagenic activities of carcinogens. Cancer Letters 11,29-33.
    Ayer, A., Tan, S.X., Grant, C.M., Meyer, A.J., Dawes, I.W., Perrone, G.G., 2010. The critical role of glutathione in maintenance of the mitochondrial genome. Free Radical Biology and Medicine 49,1956-1968.
    Azqueta, A., Arbillaga, L., Pachon, G., Cascante, M., Creppy, E.E., Lopez de Cerain, A.,2007. A quinoxaline 1,4-di-N-oxide derivative induces DNA oxidative damage not attenuated by vitamin C and E treatment. Chemico Biological Interaction 168,95-105.
    Baars, A.J., van der Molen, E.J., Spierenburg, T.J., de Graaf, G.J., Nabuurs, M.J.A., Jager, L.P.,1988. Comparative toxicity of three quinoxaline-di-N-dioxide feed additives in young pigs. Archives of Toxicology.12,405-409.
    Barendsz, A.W.,1998. Food safety and total quality management. Food Control 9 (2-3),163-170.
    Barlow, S.M., Greig, J.B., Bridges, J.W., Carere, A., Carpy, A.J.M., Galli, C.L., Kleiner, J., Knudsen, I., Koeter, H.B.W.M., Levy, L.S., Madsen, C. Mayer, S., Narbonne, J.F., Pfannkuch, F., Prodanchuk, M.G., Smith, M.R. Steinberg, P.,2002. Hazard identification by methods of animal-based toxicology. Food and Chemical Toxicology 40,145-191.
    Bauche, F., Fouchard, M.H., Jegou, B.,1994. Antioxidant system in rat testicular cells. FEBS Letters 349,392-396.
    Bedello, P.G., Goitre, M., Cane, D., Roncarolo, G.,1985. Allergic contact dermatitis to Bayo-N-OX-1. Contact Dermatitis.12,284.
    Belhadjali, H., Marguerry, M.C., Journe, F., Giordano-Labadie, F., Lefevre, H., Bazex, J.,2002. Allergic and photoallergic contact dermatitis to Olaquindox in a pig breeder with prolonged photosensitivity. Photodermatology, Photoimmunology and Photomedicine 18,52-53.\
    Bernhardt, R.,1996. Cytochrome P450:structure, function, and generation of reactive oxygen. Reviews of Physiology, Biochemistry & Pharmacology 127, 180-195.
    Beutin, L., Preller, E., Kowalski, B.,1981. Mutagenicity of quindoxin, its metabolites, and two substituted quinoxaline-di-N-oxides. Antimicrobial Agents and Chemotheropy 20,336-343.
    Borras, C., Sastre, J., Garcia-Sala, D., Lloret, A., Pallardo Federico, V., Vina, J.,2003. Mitochondria from females exhibit higher antioxidant gene expression and lower oxidative damage than males. Free Radical Biology and Medicine 34,546-552.
    Boujbiha, M.A., Hamden, K., Guermazi, F., Bouslama, A., Omezzine, A. Kammoun, A., Feki, A.E.,2009. Testicular toxicity in mercuric chloride treated rats:Association with oxidative stress. Reproductive Toxicology 28,81-89.
    Bruce, R.D.,1985. An up and down procedure for acute toxicity testing. Toxicological Science 5,151-157.
    Bruce, R.D.,1987. A confirmatory study of the up and down method for acute oral toxicity testing. Fundamental and Applied Toxicology 8,97-100.
    Burke, D.A., Wedd, D.J., Burlinson, B.,1996. Use of the Miniscreen assay to screen novel compounds for bacterial mutagenicity in the pharmaceutical industry. Mutagenesis 11,201-205.
    Carta, A., Corona, P., Loriga, M.,2005. Quinoxaline 1,4-dioxide:a versatile scaffold endowed with manifold activities. Current Medicinal Chemistry 12, 2259-2272.
    Cerna, M., Angelis, K.,1986. Studies on the mutagenic action of the quinoxaline-type growth stimulants on microorganisms. Biologizace a Chemizace Zivocisne Vyroby-Veterinaria 22,9-16.
    Chen, Q., Chen, Y., Qi, Y., Hao, L., Tang, S., Xiao, X.,2008. Characterization of carbadox-induced mutagenesis using a shuttle vector pSP189 in mammalian cells. Mutation Research 638,11-16.
    Chen, Q., Tang, S., Jin, X., Zou, J., Chen, K., Zhang, T., Xiao, X.,2009. Investigation of the genotoxicity of quinocetone, carbadox and olaquindox in vitro using Vero cells. Food and Chemical Toxicology 47,328-334.
    Chowdhury, G., Delshanee, K., Daniels, J.S., Barnes, C.H., Gates, K.S., 2004. Enzyme-activated, hypoxia-selective DNA damage by 3-amino-2-quinoxalinecarbonitrile 1,4-di-N-oxide. Chemical Research Toxicology 17, 1399-1405.
    Chun, C.,2005. Careful use of mequindox in chicken. China poultry 04. (Chinese article).
    Cihak, R., Srb, V.,1983b. Cytogenetic effects of quinoxaline-1,4-dioxide-type growth promoting agents. Ⅰ. Micronucleus test in rats. Mutation research 116,129-135.
    Cihak, R., Vontorkova, M.,1983a. Cytogenetic effects of quinoxaline-1,4-dioxide-type growth-promoting agents. Ⅱ. Metaphase analysis in mice. Mutation research 117,311-316.
    Cihak, R., Vontorkova, M.,1985. Cytogenetic effects of quinoxaline-1,4-dioxide-type growth-promoting agents. Ⅲ. Transplacental micronucleus test in mice. Mutation research 144,81-84.
    Clark, B.J., Soo, S.C., Caron, K.M., Ikeda, Y., Parker, K.L., Stocco, D.M., 1995. Hormonal and developmental regulation of the steroidogenic acute regulatory protein. Molcular Endocrinology 9,1346-1355.
    Cohn, J.N., Colucci, W.,2006. Cardiovascular effects of aldosterone and post-acute myocardial infarction pathophysiology. American Journal of Cardiology 10,4-12.
    Colby, H.D., Longhurst, P.A.,1997. Toxicology of the adrenal gland. In: Thomas, J.A., Colby, H.D. (Eds.), Endocrine Toxicology,2nd edn. Taylor & Francis, London, pp.243-281.
    Connell, J.M., Davies, E.,2005. The new biology of aldosterone. Journal of Endocrinology 186,1-20.
    Corpet, D.E.,1996. Microbiological hazards for humans of antimicrobial growth promoter use in animal production. Rev. de Med. Vet.147,851-865.
    Craigmill, A.L., Cortright, K.A.,2002. Interspecies Considerations in the Evaluation of Human Food Safety for Veterinary Drugs. AAPS Pharmacology Science 4 (4):article 34.
    Creasy, D.M.,2001a. Histopathology of the Male Reproductive System Ⅰ: Techniques. John Wiley & Sons, Inc.
    Creasy, D.M.,2001b. Histopathology of the Male Reproductive System Ⅱ: Interpretation. John Wiley & Sons, Inc.
    Creasy, D.M.,2001c. Pathogenesis of male reproductive toxicity. Toxicologic Pathology 29,64-76.
    Dafa, Li.,2004. Use of alternatives to AGP in animal food production in China. Working papers from the international symposium:DIAS report Animal Husbandry no.57.
    Dawson, A.B.,1926. A note on the staining of the skeleton of cleared specimens with Alizarin. Red S. Stain Technology 1,123-124.
    De Vries, H., Beyersbergen van Henegouwen, G.M., Kalloe, F., Berkhuysen, M.H.,1990a. Phototoxicity of olaquindox in the rat. Research in Veterinary Science 48,240-244.
    De Vries, H., Bojarski, J., Donker, A.A., Bakri, A., Beyersbergen van Henegouwen, G.M.,1990b. Photochemical reactions of quindoxin, olaquindox, carbadox and cyadox with protein, indicating photoallergic properties. Toxicology 63,85-95.
    DECOS,1999. Health Council of the Netherlands, Dutch Expert Committee on Occupational Standards. Carbadox. The Hague:Health Council of the Netherlands, pub No.1999/06OSH
    Diemer, T., Allen, J.A., Hales, K.H., Hales, D.B.,2003. Reactive oxygen disrupts mitochondria in MA-10 tumor Leydig cells and inhibits steroidogenic acute regulatory (StAR) protein and steroidogenesis. Endocrinology 144, 2882-2891.
    Doreswamy, K., Muralidhara,2005. Genotoxic consequences associated with oxidative damage in testis of mice subjected to iron intoxication. Toxicology 206,169-178.
    Droge, W.,2002. Free radicals in the physiological control of cell function. Physiological Review 82,47-95.
    Droumev, D.,1983. Review of antimicrobial growth promoting agents available. Veterinary Research Communications 7,85-99.
    Dybing, E., Doe, J., Groten, J., Kleiner, J., O'Brien, J., Renwick, A.G., Schlatter, J., Steinberg, P., Tritscher, A., Walker, R., Younes, M.,2002. Hazard characterisation of chemicals in food and diet:dose response, mechanisms and extrapolation issues. Food and Chemical Toxicology 40, 237-282.
    England, K., Cotter, T.G.,2005. Direct oxidative modifications of signalling proteins in mammalian cells and their effects on apoptosis. Redox Report 10, 237-245.
    EU,1998. Commission Regulation No.2788/98, Off. J. Eur. Community L347,3.
    Fang, G.,2006. Preclinical toxicological safety evaluation of cyadox. PhD thesis-Huazhong Agriculture University, Wuhan.
    Fang, G., He, Q., Zhou, S., Wang, D., Zhang, Y., Yuan, Z.,2006. Subchronic oral toxicity study with cyadox in Wistar rats. Food and Chemical Toxicology 44,36-41.
    FDA (United States Food and Drug Administration).2005. Guidance for Industry #3. FDA General Principles for Evaluating the Safety of Compounds Used in Food-producing Animals.
    FDA,1998. Freedom of information summery (MECADOX〔R〕 10); NADA 041-061. http://www.fda.qov/cvm/FOI/520.htm.
    FDA,2000a. Toxicological Principles for the Safety Assessment of Food Ingredients. IV.C.9.a. Guidelines for Reproduction Toxicity Studies.
    FDA,2000b. Toxicological principles for the safety assessment of food ingredients. IV.C.1.a.Bacterial Reverse Mutation Test.
    FDA,2000c. Toxicological principles for the safety assessment of food ingredients. IV.C.1.b. Mammalian Chromosomal Aberration Test.
    FDA,2000d. Toxicological principles for the safety assessment of food ingredients. IV.C.1.d. Mammalian Erythrocyte Micronucleus Test.
    Feng, W.,2008. Investigation of mequindox for the antibacterial activity on common pathogenic bacteria of livestock and poultry in vitro. Contemporary Animal Husbandry 6,47-49.
    Ferrando, R., Truhaut, R., Raynaud, J.P.,1977. Principles of a full'relay toxicity' experiment and results conducted with carbadox, a feed additive used as growth promoter for growing swine. Folia Veterinaria Latina 7,333-340.
    Frain-Bell, W., Gardiner, J.,1975. Photocontact dermatitis due to quindoxin. Contact Dermatitis 1,256-257.
    Francalanci, S., Gola, M., Giorgini, S., Muccinelli, A., Sertoli, A.,1986. Occupational photocontact dermatitis from Olaquindox. Contact Dermatitis 15, 112-114.
    Gad, S.C., Chengelis, C.P.,1998. Routes, Formulations, and Vehicles. In: Acute Toxicology Testing (Second Edition). Academic Press, San Diego, pp. 297.
    Garcia-Franco, S., Dominguez, G., Pico, J.C.,1999. Alternatives in the induction and preparation of phenobarbital/naphthoflavone-induced S9 and their activation profiles. Mutagenesis 14,323-326.
    Graaf, C., Spierenburg, T., Baars, A.,1990. Proceedings of the EuroResidue I, Noord-wijkerhout, The Netherlands, pp.202-205.
    Groves, B.I., Flipot, P.M., Hartsock, T.G., Vlielander, L.C.,1983. The effect of carbadox on reproduction when fed to Swine up to 60 kilograms body weight. Canadian Veterinary Journal 24,154-157.
    Gu, W., Hecht, N.B.,1996. Developmental expression of glutathione peroxidase, catalase, and manganese superoxide dismutase mRNAs during spermatogenesis in the mouse. Journal of Andrology 17,256-262.
    Guyton, A.C. and Hall, J.E.,2006. Adrenocortical Hormones. In Textbook of Medical Physiology, Elsevier, Saunders, Philadelphia, pp.957.
    Halliwell, B., Gutteridge, J.M.,1984. Lipid peroxidation, oxygen radicals, cell damage, and antioxidant therapy. Lancet 323,1396-1397.
    Hao, L., Chen, Q., Xiao, X.,2006. Molecular mechanism of mutagenesis induced by olaquindox using a shuttle vector pSP189/mammalian cell system. Mutation Research 599,21-25.
    Hayashi, M., Norppa, H., Sofuni, T., Ishidate, M., Jr.,1992. Mouse bone marrow micronucleus test using flow cytometry. Mutagenesis 7,251-256.
    He, Q., Fang, G., Wang, Y., Wei, Z., Wang, D., Zhou, S., Fan, S., Yuan, Z., 2006. Experimental evaluation of cyadox phototoxicity to Balb/c mouse skin. Photodermatology, Photoimmunology and Photomedicine 22,100-104.
    Hinson, J.P., Raven, P.W.,2006. Effects of endocrine-disrupting chemicals on adrenal function. Best Practice & Research Clinical Endocrinology & Metabolism 20,111.
    Hongxia, L., Wei J., Guilan Z.,2005. Mequindox poisioning; diagnosis and treatment. Livestock and Poultry Industry 4,54-55 (Chinese article).
    Hu, W., Xie, J., Zhao, J., Xu, Y., Yang, S., Ni, W.,2009. Involvement of Bcl-2 family in apoptosis and signal pathways induced by cigarette smoke extract in the human airway smooth muscle cells. DNA and Cell Biology 1,13-22.
    Huang, X.J., Ihsan, A., Wang, X., Dai, M.H., Wang, Y.L., Su, S.J., Xue, X.J. Yuan, Z.H.,2009. Long-term dose-dependent response of Mequindox on aldosterone, corticosterone and five steroidogenic enzyme mRNAs in the adrenal of male rats. Toxicology Letters 191,167-173.
    Huang, X.J., Wang, X., Ihsan, A., Liu, Q., Xue, X.J., Su, S.J., Yang, C.H. Zhou, W., Yuan, Z.H.,2010a. Interactions of NADPH oxidase, renin-angiotensin-aldosterone system and reactive oxygen species in mequindox-mediated aldosterone secretion in Wistar rats. Toxicology Letters 198,112-118.
    Huang, X.J., Zhang, H.H., Wang, X., Huang, L.L., Zhang, L.Y., Yan, C.X., Liu, Y., Yuan, Z.H.,2010b. ROS mediated cytotoxicity of porcine adrenocortical cells induced by QdNOs derivatives in vitro. Chemico-Biological Interactions 185,227-234.
    Ihsan, A., Wang, X., Huang, X.J., Liu, Y., Liu, Q., Zhou, W., Yuan, Z.H. 2010. Acute and subchronic toxicological evaluation of Mequindox in Wistar rats. Regulatory Toxicology and Pharmacology 57,307-314.
    IPCS,1987, Principles for the safety assessment of food additives and contaminants in food. WHO Environmental Health Criteria No.70. International Programme on Chemical Safety (Geneva:World Health Organization).
    Jager, L.P., De Graaf, G.J., Widjaja-Greefkes, H.C., Accord Burleson, C.C., Van den Dungen, H.M., Baars, A.J.,1994. Effects of feed additives and veterinary drugs on aldosterone production and release by porcine adrenal cells in vitro. Journal of Veterinary Pharmacology and Theraputics 17,175-185.
    Jager, L.P., De Graaf, G.J., Widjaja-Greefkes, H.C.A.,1996. Screening for drug induced alterations in the production and release of steroid hormones by porcine adrenocortical cells in vitro. Toxicology in Vitro 10,595-608.
    Jager, L.P., Vroomen, L.H.,1990. [Toxicological considerations in the evaluation of veterinary drugs]. Tijdschr Diergeneeskd 115,727-735.
    Janer, G., Hakkert, B.C., Piersma, A.H., Vermeire, T., Slob, W.,2007. A retrospective analysis of the added value of the rat two-generation reproductive toxicity study versus the rat subchronic toxicity study. Reproductive Toxicology 24,103-113.
    Kackar, R., Srivastava, M.K., Raizada, R.B.,1997. Induction of gonadal toxicity to male rats after chronic exposure to mancozeb. Industrial Health 35, 104-111.
    Kheradmand, A., Alirezaei, M., Asadian, P., Rafiei Alavi, E., Joorabi, S., 2009. Antioxidant enzyme activity and MDA level in the rat testis following chronic administration of ghrelin. Andrologia 41,335-340.
    Kinoshita, A., Wanibuchi, H., Imaoka, S., Ogawa, M., Masuda, C., Morimura, K., Funae, Y., Fukushima, S.,2002. Formation of 8-hydroxydeoxyguanosine and cell-cycle arrest in the rat liver via generation of oxidative stress by phenobarbital:association with expression profiles of p21WAF1/Cip1, cyclin D1 and Ogg1. Carcinogenesis 23,341-349.
    Krishna, G. and Makoto, H.,2000. In vivo rodent micronucleus assay: protocol, conduct and data interpretation. Mutation Research 155-166.
    Kumar, S., Sitasawad, S.L.,2009. N-acetylcysteine prevents glucose/glucose oxidase induced oxidative stress, mitochondrial damage and apoptosis inH9c2 cells. Life Science 84,328-336.
    Kumar, T.R., Muralidhara,2007. Induction of oxidative stress by organic hydroperoxides in testis and epididymal sperm of rats in vivo. Journal of Andrology 28,77-85.
    Lai, A., Veinot, J.P., Ganten, D., Leenen, F.H.H.,2005. Prevention of cardiac remodeling after myocardial infarction in transgenic rats deficient in brain angiotensinogen. Journal of Molcular and Cellular Cardiology 39,521-529.
    Lee, E., Ahn, M.Y., Kim, H.J., Kim, I.Y., Han, S.Y., Kang, T.S., Hong, J.H., Park, K.L., Lee, B.M., Kim, H.S.,2007. Effect of di(n-butyl) phthalate on testicular oxidative damage and antioxidant enzymes in hyperthyroid rats. Environmental Toxicology 22,245-255.
    Li,2006. Common poisoning and prevention in rural chicken. Veterinary drugs & feed additives 06 (Chinese article).
    Li, P., Nijhawan, D., Budihardjo, I., Srinivasula, S.M., Ahmad,M., Alnemri, E.S.,Wang, X.,1997. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 91,47-489.
    Lilienblum, W., Dekant, W., Foth, H., Gebel, T., Hengstler, J., Kahl, R., Kramer, P.J., Schweinfurth, H., Wollin, K.M.,2008. Alternative methods to safety studies in experimental animals:role in the risk assessment of chemicals under the new European Chemicals Legislation (REACH). Archives of Toxicology 82,211-236.
    Lisurek, M., Bernhardt, R.,2004. Modulation of aldosterone and cortisol synthesis on the molecular level. Molecular and Cellular Endocrinology 25, 149-159.
    Liu, Z., Huang, L., Dai, M., Chen, D., Tao, Y., Wang, Y., Yuan, Z.,2009. Metabolism of cyadox in rat, chicken and pig liver microsomes and identification of metabolites by accurate mass measurements using electrospray ionization hybrid ion trap/time-of-flight mass spectrometry. Rapid Communications in Mass Spectrometry 23,2026-2034.
    Liu, Z., Huang, L., Dai, M., Chen, D., Wang, Y., Tao, Y., Yuan, Z.,2008. Metabolism of olaquindox in rat liver microsomes:structural elucidation of metabolites by high-performance liquid chromatography combined with ion trap/time-of-flight mass spectrometry. Rapid Communications in Mass Spectrometry 22,1009-1016.
    Liu, Z.Y., Huang, L.L., Chen, D.M., Dai, M.H., Tao, Y.F., Yuan, Z.H.,2010a. The metabolism and N-oxide reduction of olaquindox in liver preparations of rats, pigs and chicken. Toxicology Letters 195,51-59.
    Liu, Z.Y., Huang, L.L., Chen, D.M., Yuan, Z.H.,2010b. Metabolism of mequindox in liver microsomes of rats, chicken and pigs. Rapid Communications in Mass Spectrometry 24,909-918.
    Livingstone, D.R.,2001. Contaminant-stimulated reactive oxygen species production and oxidative damage in aquatic organisms. Marine Pollution Bulletin 42,656-666.
    Lu, F.C.,1985. Acute, short-term and long-term toxicity studies. In:Basic Toxicology:Fundamentals, Target Organs, and Risk Assessment, pp.99-104.
    Markovic, B., Stanimirovic, Z., Bajic, V.,2000a. The genotoxicity of Carbadox in vitro. Abstracts from EEMS-2000 (30th Annual Meeting of the European Environmental Mutagen Society) 86,105.
    Markovic, B., Stanimirovic, Z., Vucinic, M., Cupic, V.,2000b. Examination of Carbadox genotoxicity in vitro and in vivo. Acta.Veterinaria (Belgrade) 50, 387-396.
    Mehta, A.K., Arora, N., Gaur, S.N., Singh, B.P.,2009. Acute toxicity assessment of choline by inhalation, intraperitoneal and oral routes in Balb/c mice. Regulatory Toxicology and Pharmacology 54,282-286.
    Mejia, L.A., Korgaonkar, C.K., Schweizer, M., Chengelis, C., Novilla, M., Ziemer, E., Williamson-Hughes, P.S., Grabiel, R., Empie, M.,2009. A 13-week dietary toxicity study in rats of a Napin-Rich Canola Protein Isolate. Regulatory Toxicology and Pharmacology 55,394-402.
    Monari, M., Foschi, J., Matozzo, V., Marin, M.G., Fabbri, M., Rosmini, R., Serrazanetti, G.P.,2009. Investigation of EROD, CYP1A immunopositive proteins and SOD in haemocytes of Chamelea gallina and their role in response to B[a]P. Comparative Biochemistry and Physiology. Part C: Toxicology and Pharmacology 149,382-392.
    Mortelmans, K., Zeiger, E.,2000. The Ames Salmonella/microsome mutagenicity assay. Mutatation Research 455,29-60.
    Muirhead, S.,1998. EU ban of antibiotics draws sharp criticism. Feedstuffs 70,1.
    Murphy, M.P.,2009. How mitochondria produce reactive oxygen species. Biochemical Journal 417,1-13.
    Nabuurs, M.J., van der Molen, E.J.,1989. Clinical signs and performance of pigs during the administration of different levels of carbadox and after withdrawal. Zentralbl Veterinarmed A 36,209-217.
    Nabuurs, M.J., van der Molen, E.J., de Graaf, G.J., Jager, L.P.,1990. Clinical signs and performance of pigs treated with different doses of carbadox, cyadox and olaquindox. Zentralbl Veterinarmed A 37,68-76.
    Nastuneak, J., Kolouch, F., Vanova, L., Dvorak, M., Sevick, B.,1986. Three month toxicity to laboratory Norway rats:Cyadox compared with olaquindox. Bio. Chem. Vet. (Prague) 22,367-380.
    Negishi, T., Tanaka, K., Hayatsu, H.,1980. Mutagenicity of carbadox and several quinoxaline 1,4-dioxide derivatives. Chemical and Pharmaceutical Bulletin (Tokyo) 28,1347-1349.
    Nestmann, E.R., Lynch, B.S.,2007. Method for calculating ADI-derived guidance values for drug carryover levels in medicated feeds. Regulatory Toxicology and Pharmacology 47,232-239.
    Newsholme, S.J., Walton, J.R., Elliott, G.,1986. Poor growth, episodic paralysis and adrenocortical injury in swine after accidental olaquindox overdosage. Veterinary Record 29,554-555.
    Nilsson, R.,2000. Endocrine modulators in the food chain and environment. Toxicologic Pathology 28,420-431.
    Nishimura, K.A.,1974. Microdissection method for detecting thoracic visceral malformations in mouse and rat fetuses. Congenital Anomalies14,23-40.
    Novave, K.L., Semenkova, L., Holik, M.,1986. Deoxidation of quinoxaline-1,4-dioxide derivative in the presence of aminoacids. Cesk Farm 35,145-150.
    NRC,2004. The Development of Science based Guidelines for Laboratory Animal Care, Proceedings of the November 2003 International Workshop, National Academy Press, Washington, DC.
    Nunoshiba, T., Nishioka, H.,1989. Genotoxicity of quinoxaline 1,4-dioxide derivatives in Escherichia coli and Salmonella typhimurium. Mutation Research 217,203-209.
    OECD,2001a. Test Guideline 425, Acute Oral Toxicity-"Up and Down Procedure", adopted 17th, December.
    OECD,2001b. Test Guideline 420, Acute Oral Toxicity-"Fixed Dose Procedure", adopted 17th, December.
    Ohta, T., Moriya, M., Kaneda, Y., Watanabe, K., Miyazawa, T., Sugiyama, F., Shirasu, Y.,1980. Mutagenicity screening of feed additives in the microbial system. Mutation Research 77,21-30.
    Ong, C.N., Shen, H.M., Chia, S.E.,2002. Biomarkers for Male Reproductive health hazards:Are they available? Toxicology Letters 134,17-30.
    Oud, J.L., Reutlinger, A.H., Branger, J.,1979. An investigation into the cytogenetic damage induced by the coccidiostatic agents amprolium, carbadox, dimetridazole and ronidazole. Mutation Research 68,179-182.
    Peltola, V., Huhtaniemi, I., Ahotupa, M.,1992. Antioxidant enzyme activity in the maturing rat testis. Journal of Andrology 13,450-455.
    Pokorna, D.,1986. Cytogentic analysis of bone marrow cells in Chinese hamster after the administration of cyadox and olaquindox. Biol. Chem. Vet. (Prague) 22,23-28.
    Power, S.B., Donnelly, W.J., McLaughlin, J.G., Walsh, M.C., Dromey, M.F. 1989. Accidental carbadox overdosage in pigs in an Irish weaner-producing herd. Veterinary.Record 124,367-370.
    Prieto, P., Baird, A.W., Blaauboer, B.J., Castell Ripoll, J.V., Corvi, R., Dekant, W., Dietl, P., Gennari, A., Gribaldo, L., Griffin, J.L., Hartung, T. Heindel, J.J., Hoet, P., Jennings, P., Marocchio, L., Noraberg, J., Pazos, P., Westmoreland, C., Wolf, A., Wright, J., Pfaller, W.,2006. The assessment of repeated dose toxicity in vitro:a proposed approach. The report and recommendations of ECVAM workshop 56. Alternative Laboratory Animals 34, 315-341.
    Priyadarsini, K.I., Dennis, M.F., Naylor, M.A., Stratford, M.R.L., Wardman, P.,1996. Free radical intermediates in the reduction of quinoxaline N-oxide antitumor drugs:redox and prototropic reactions. Journal of American Chemical Society 118,5648-5654.
    Raven, P.W., Hinson, J.P.,1996. Transport, actions and metabolism of adrenal hormones and pathology and pharmacology of the adrenal gland. In: Harvey, P.W. (Ed.), The Adrenal in Toxicology:Target Organ andModulator of Toxicity. Taylor and Francis, London, pp.53-79.
    Raza, H., John, A., Brown, E.M., Benedict, S., Kambal, A.,2008. Alterations in mitochondrial respiratory functions, redox metabolism and apoptosis by oxidant 4-hydroxynonenal and antioxidants curcumin andmelatonin in PC12 cells. Toxicology and Applied Pharmacology 226,161-168.
    Redfern, B. G., Wise, L. D.,2007. High-throughput staining for the evaluation of fetal skeletal development in rats and rabbits. Birth Defects Research Part B Developmental and Reproductive Toxicology.80(3),177-182.
    Rispin, A., Farrar, D., Margosches, E., Gupta, K., Stitzel, K., Carr, G., Greene, M., Meyer, W., McCall, D.,2002. Alternative methods for the median lethal dose (LD50) test:the up-and-down procedure for acute oral toxicity. ILAR Journal 43,233-243.
    Rosol, T.J., Yarrington, J.T., Latendresse, J., Capen, C.C.,2001. Adrenal gland:structure, function, and mechanisms of toxicity. Toxicologic Pathology 29,41-48.
    Sanchez-Perez, J., Lopez, M.P.& Garcia-Diez, A.,2002. Airborne allergic contact dermatitis from olaquindox in a rabbit breeder. Contact Dermatitis 46, 185.
    SCAN,1998. Opinion of the Scientific Committee for Animal Nutrition on possible risks for the consumer, the animal and the users from the use of Carbadox and Olaquindox as Feed Additives,10 July. www.bmgfj.gv.at/cms/site/attachments /6/2/4/CH0264/CMS1085743089437/opinion1 bt176.pdf
    Scheutwinkel-Reich, M., vd Hude, W.,1984. Sister-chromatid exchange in Chinese hamster V79 cells exposed to quindoxin, carbadox and olaquindox. Mutation Research 139,199-202.
    Schlottmann, K., Scholmerich, J.,1999. Bcl-2 family-members andmitochondria. In:Schunkert, H., Riegger, G. (Eds), Apoptosis in Cardiac Biology. Kluwer Academic Publishers, Boston, pp.71-90.
    Schmidt, B.M., Ilic, N., Poulev, A., Raskin, I.,2007. Toxicological evaluation of a chicory root extract. Food and Chemical Toxicology 45,1131-1139.
    Scott, K.W., Dawson, T.A.,1974. Photo-contact dermatitis arising from the presence of quindoxin in animal feeding stuffs. British Journal of Dermatology 90,543-546.
    Sevcik, B., et al.,1986. Research of harmless and safety of application of the growth promoter cyadox. Workshop 1,93.
    Shi, Z., Zhang, H., Liu, Y., Xu, M., Dai, J.,2007. Alterations in gene expression and testosterone synthesis in the testes of male rats exposed to perfluorododecanoic acid. Toxicological Science 98,206-215.
    Sies, H.,1997. Oxidative stress:oxidants and antioxidants. Experimental Physiology 82,291-295.
    Sikka, S.C.,1996. Oxidative stress and role of antioxidants in normal and abnormal sperm function. Frontiers in Bioscience1,78-86.
    Sikka, S.C.,2004. Role of oxidative stress and antioxidants in andrology and assisted reproductive technology. Journal of Andrology 25,5-18.
    Smith, G.C.M., Tew, D.G., Wolf, C.R.,1994. Dissection of NADPH-cytochrome P450 oxidoreductase into distinct functional domains. Proceedings of the National Academy of Sciences 91,8710-8714.
    Snyder, M.J.,2000. Cytochrome P450 enzymes in aquatic invertebrates: recent advances and future directions. Aquatic Toxicology 48,529-547.
    Spierenburg, T.J., Baars, A.J., de Graaf, G.J., Jager, L.P.,1988. Carbadox-induced inhibition of aldosterone production in porcine adrenals in vitro. Toxicology in Vitro 2,141-143.
    Spierenburg, T.J., van der Holen, E.J., de Graaf, G.J., Jager, L.P., Baars, A.J.,1989. Adrenal toxicity of carbadox, olaquindox and cyadox in weaned piglets. Abstracts from Toxicological Meeting. Pharmacy World & Science.11, B9.
    Sram, R.J., Fernandez, R.S.F., Kocisova, J.,1986a. Chromosomal aberration in the bone marrow of mice after long-continued administration cyadox and olaquindox. Biol. Chem. Vet. (Prague) 22,17-22.
    Sram, R.J., Fernandez, R S.F., Kocisova, J.,1986b. Effect of long-term oral administration of cyadox and olaquindox on the frequency of dominant lethal mutations and sperm abnormalities in mice. Biol. Chem. Vet. (Prague) 22,37-45.
    Sram, R.J., Fernandez, R.S.F., Kocisova, J.,1986c. Dominant lethal mutation in female mice following the oral application of cyadox and olaquindox. Bio. Chem. Vet. (Prague) 22,29-35.
    Stocco, D.M.,1998. A review of the characteristics of the protein required for the acute regulation of steroid hormone biosynthesis:the case for the steroidogenic acute regulatory (StAR) protein. Proceedings of the Society for Experimental Biology and Medicine 217,123-129.
    Stocco, D.M.,2001. StAR protein and the regulation of steroid hormone biosynthesis. Annual Review of Physiology 63,193-213.
    Suter, W., Rosselet, A.,& Knusel, F.,1978. Mode of action of quindoxin and substituted quinoxaline-di-N-oxides on Escherichia coli. Antimicrobial Agents Chemotherapy 13,770-783.
    Swierenga, S.H., Heddle, J.A., Sigal, E.A., Gilman, J.P., Brillinger, R.L. Douglas, G.R., Nestmann, E.R.,1991. Recommended protocols based on a survey of current practice in genotoxicity testing laboratories, IV. Chromosome aberration and sister-chromatid exchange in Chinese hamster ovary, V79 Chinese hamster lung and human lymphocyte cultures. Mutation Research 246,301-322.
    Sykora, I.,1977. Effect of non-antibiotic stimulators on laboratory rats. Veterinary Medicine (Praha) 22,377-384.
    Sykora, I., Marhan, O., Gandalovicova, D.,1981. [The effect of the non-antibiotic growth stimulators, Carbadox and Cyadox, on reproduction in laboratory animals]. Veterinary Medicine (Praha) 26,367-377.
    Sykora, I., Vortel V.,1986. Post-natal study of the carcinogenicity of cyadox and carbadox. Biol. Vet (Praha) 22,52-62.
    Tahka, K.M.,1989. Local control mechanisms in the testis. Inernationalt Journal of Developmental Biology 33,141-148.
    Tokosova, M.,1989. The effect of growth stimulators on the levels of various macroelements in chickens. Veterinary Medicine (Praha) 34(1),51-57.
    Toyokuni, S.,1999. Reactive oxygen species-induced molecular damage and its application in pathology. Pathology Internationa149,91-102.
    Truhaut, R., Ferrando, R., Faccini, J.M., Monro, A.M.,1981. Negative results of carcinogenicity bioassay of methyl carbazate in rats:significance for the toxicological evaluation of carbadox. Toxicology22,219-221.
    Tu, Honggang,2007. The genotoxicity of quinoxalines to mammalian cells. M.Sc. Thesis-Huazhong Agriculture University, Wuhan.
    Tucker, M.J.,1975. Carcinogenic action of quinoxaline 1,4-dioxide in rats. Journal of the National Cancer Institute 55,137-146.
    Turner, T.T., Lysiak, J.J.,2008. Oxidative Stress:A Common Factor in Testicular Dysfunction. Journal of Andrology 29,488-498.
    Ungemach, F.R.,1995. Safety aspects of growth promoters used as feed additives. In:Proceedings of the Scientific Conference on Growth Promotion in Meat Production, vol.29, Brussels, Belgium, pp.333-346.
    Van de Kerk, P.,1985. Drug toxicity in piglets following long term consumption of medicated feed. Tijdschr Diergeneeskd 110,181-184.
    Van der Molen, E.J.,1988. Pathological effects of carbadox in pigs with special emphasis on the adrenal. Journal of Comparative Pathology 98,55-67.
    Van der Molen, E.J., Baars, A.J., de Graaf, G.J., Jager, L.P.,1989a. Comparative study of the effect of the effect of carbadox, olaquindox and cyadox on aldosterone, sodium and potassium plasma levels in weaned pigs. Research in Veterinary Science 47,11-16.
    Van der Molen, E.J., de Graaf, G.J., Baars, A.J.,1989b. Persistence of carbadox induced adrenal lesions in pigs following drug withdrawal and recovery of aldosterone plasma concentrations. Journal of Comparative Pathology 100,295-304.
    Van der Molen, E.J., de Graaf, G.J., Baars, A.J., Schopman, W.,1986a. Carbadox induced changes in aldosterone, sodium and potassium levels in the blood of weaned pigs. Zentralbl Veterinarmed A 33,617-623.
    Van der Molen, E.J., de Graaf, G.J., Spierenburg, T.J., Nabuurs, M.J. Baars, A.J., Jager, L.P.,1986b. Hypoaldosteronism in piglets induced by carbadox. Experientia 42,1247-1249.
    Van der Molen, E.J., Nabuurs, M.J., Jager, L.P.,1985. Pathological and clinical changes related to toxicity of carbadox in weaned pigs. Zentralbl Veterinarmed A 32,540-550.
    Van der Molen, E.J., van Lieshout, J.H., Nabuurs, M.J., Derkx, F.H., Lamers, A.P., Michelakis, A.M.,1989c. Changes in plasma renin activity and renal immunohistochemically demonstrated renin in carbadox treated pigs. Research in Veterinary Science 46,401-405.
    Van Miert, A.S., Vroomen, L.H., Jager, L.P.,1984. [Pharmacology and toxicology of furazolidone and carbadox; a literature study]. Tijdschr Diergeneeskd 109,928-933.
    Van Schie, F.W.,1982. [A case of carbadox poisoning]. Tijdschr Diergeneeskd 107,428.
    Vanova, L., Kolouch, F., Sevcik, B., Strakova, J.,1986. The action of cyadox on the reproduction of rabbit. Biologizace a Chemizace Zivocisne Vyroby-Veterinaria 22,215-222.
    Vicente, E., Perez-Silanes, S., Lima, L.M., Ancizu, S., Burguete, A., Solano, B., Villar, R., Aldana, I., Monge, A.,2009. Selective activity against Mycobacteriumtuberculosis of new quinoxaline 1,4-di-N-oxides. Bioorganic and Medicinal Chemistry 17,385-389.
    Von der Hude, W., Behm, C., Gurtler, R., Basler, A.,1988. Evaluation of the SOS chromotest. Mutation Research 203,81-94.
    Voogd, C.E., van der Stel, J.J., Jacobs, J.J.,1980. The mutagenic action of quindoxin, carbadox, olaquindox and some other N-oxides on bacteria and yeast. Mutation Research 78,233-242.
    Waldmann, K.H., Kikovic, D., Stockhofe, N.,1989. Clinical and hematological changes after olaquindox poisoning in fattening pigs. Zentralbl Veterinarmed A 36,676-686.
    Walker, R.,1998. Toxicity testing and derivation of the ADI. Food Additives and Contaminants 15, Supplement,11-16.
    Walker, R.,1999. The Significance of Excursions above the ADI:Duration in Relation to Pivotal Studies. Regulatory Toxicology and Pharmacology 30, 114-118.
    Wang, X., Fang, G.J., Wang, Y.L., Ihsan, A., Huang, L.L., Zhou, W., Liu, Z.L., Yuan, Z.H.,2011. Two generation reproduction and teratogenicity studies of feeding cyadox in Wistar rats. Food and Chemical Toxicology 49, 1068-1079.
    Wang, X., He, Q.H., Wang, Y.L., Ihsan, A., Huang, L.L., Zhou, W., Su, S.J., Liu, Z.L., Yuan, Z.H.,2010a. A chronic toxicity study of cyadox in Wistar rats. Regulatory Toxicology and Pharmacology 59,324-333.
    Wang, X., Huang, X.J., Ihsan, A., Liu, Z.Y., Huang, L.L., Zhang, H.H., Zhang, H.F., Zhou, W., Liu, Q., Xue, X.J., Yuan, Z.H.,2010b. Metabolites and JAK/STAT pathway were involved in the liver and spleen damage in male Wistar rats fed with mequindox. Toxicology 280,126-134.
    Wang, X., Zhang, W., Wang, Y., Peng, D., Ihsan, A., Huang, X., Huang, L Liu, Z., Dai, M., Zhou, W., Yuan, Z.H.,2010c. Acute and sub-chronic oral toxicological evaluations of quinocetone in Wistar rats. Regulatory Toxicology and Pharmacology 58,421-427.
    Wang, Y., Zhao, R., Xu, Z., Li, J., Yan, X., Zhou. Z.,1992a. Comparison of the effect induced by mequindox and carbadox. Chinese Journal of Veteterinary Science and Technology 22(10),29-30.
    Wang, Y.C., Yang, X.L., Zhao, R.C., Li, J.S., Xue, F.Q., Liang, J.P., Xu, Z.Z., Du, X.L 1994. Study on subacute toxicity of quinocetone in mice and rats. Journal of Traditional Chinese Vetetery Medicine 5,11.
    Wang, Y.C., Yang, X.L., Zhao, R.C., Li, J.S., Xue, F.Q., Liang, J.P., Xu, Z.Z., Du, X.L.,1992b. Study on cumulative toxicity of quinocetone in mice. Journal of Traditional Chinese Vetetery Medicine 5,14.
    Westat,2001. Acute Oral Toxicity (OECD Test Guideline 425) Statistical Programme (AOT 425 StatPgm).
    White, P.C.,2004. Aldosterone synthase deficiency and related disorders. Molecular and Cellular Endocrinology 217',81-87.
    White, R.E., Coon, M.J.,1980. Oxygen activation by cytochrome P450. Annual Review of Biochemistry 49,315-356.
    WHO (World Health Organization),1991a. Toxicological evaluation of certain veterinary drug residues in food. Carbadox. WHO Food Additives Series, No.27, no.694 on INCHEM.
    WHO (World Health Organization),1991b. Toxicological evaluation of certain veterinary drug residues in food. Olaquindox. WHO Food Additives Series, No.27, no.701 on INCHEM.
    WHO,1994. Toxicological evaluation of certain veterinary drug residues in food. WHO Food Additives Series, No.33, nos.804-817 on INCHEM.
    WHO,2003. Toxicological evaluation of certain veterinary drug residues in food. WHO Food Additives Series, No.51.
    Willianson, E.M., Okpako, D.T., Evans, F.J.,1996. Selection, Preparation and Pharmacological Evaluation of Plant Material England:John Wiley.
    Wilson, J.G.,1965. Methods for administering agents and detecting malformations in experimental animals. In:J.G. Wilson, J. Warkany (Eds.), Teratology. Principles and Techniques, University of Chicago Press, Chicago and London, pp.262-277.
    Woodward K.N.,2005a. Veterinary pharmacovigilance. Part 4. Adverse reactions in humans to veterinary medicinal products. Journal of Veterinary.Pharmacology and Theraputics 28,185-201.
    Woodward K.N.,2005b. Veterinary pharmacovigilance. Part 6. Predictability of adverse reactions in animals from laboratory toxicology studies. Journal of Veterinary.Pharmacology and Theraputics 28,213-231.
    Woodward, K.N.,2008. Assessment of user safety, exposure and risk to veterinary medicinal products in the European Union. Regulatory Toxicology and Pharmacology 50,114-128.
    Wu, Y., Yu, H., Wang, Y., Huang, L., Tao, Y., Chen, D., Peng, D., Liu, Z. Yuan, Z.,2007. Development of a high-performance liquid chromatography method for the simultaneous quantification of quinoxaline-2-carboxylic acid and methyl-3-quinoxaline-2-carboxylic acid in animal tissues. Journal of Chromatography A 1146,1-7.
    Yarrington, J.T., Latendresse, J.R., Capen, C.C.,1996. Toxic responses of the adrenal cortex. In:Sipes, I.G., McQueen, C.A., Gandol" Z, A.J. (Eds.), Comprehensive Toxicology. Elsevier Science, Oxford, pp.637-649.
    Yoshimura, H.,2002. Teratogenic assessment of carbadox in rats. Toxicology Letters 129,115-118.
    Yoshimura, H., Nakamura, M., Koeda, T., Yoshikawa, K.,1981. Mutagenicities of carbadox and olaquindox--growth promoters for pigs. Mutation Research 90,49-55.
    Zhang, W., Peng, D.P., Huang, L.L., Wang, Y.L., Yuan, Z.H.,2007. Study on the genotoxicity of quinocetone. Journal of Toxicology 21 (4),335 (Chinese article).
    Zhang, Wei,2007. Preclinical toxicology of quinocetone. M.Sc. Thesis Huazhong Agriculture University, Wuhan.
    Zhao, R., Xia, W., Wang, Y., Yan, X., Zhou, Z., Li, J., Cai, J.,1982. Initial observation of the bacteriostasis activity and toxicity of "MAQO" and "MCEQO". Journal of Traditional Chinese Vetetery Medicine 1,51-54.
    Zhen, Li.,2006. Common poisoning and prevention in rural chicken. Veterinary Pharmaceuticals & feed additives 11,30-31 (Chinese article).
    Zhu, H.L., Yuan, Z.H., Wang, Y.L., Qiu, Y.S., Fan, S.X.,2006. The effect of cyadox supplementation on metabolic hormones and epidermal growth factor in swine. Animal Science 82,350-354.
    Zorov, D.B., Bannikova, S.Y., Belousov, V.V., Vyssokikh, M.Y., Zorova, L.D., Isaev, N.K., Krasnikov, B.F., Plotnikov, E.Y.,2005. Reactive oxygen and nitrogen species:friends or foes? Biochemistry (Mosc.) 70,215-221.
    Zou, J., Chen, Q., Tang, S., Jin, X., Chen, K., Zhang, T., Xiao, X.,2009. Olaquindox induced genotoxicity and oxidative DNA damage in human hepatoma G2 (HepG2) cells. Mutation Research 676,27-33.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700